Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 4(12): 101306, 2023 12 19.
Article in English | MEDLINE | ID: mdl-38052214

ABSTRACT

Skeletal muscle atrophy is a hallmark of cachexia, a wasting condition typical of chronic pathologies, that still represents an unmet medical need. Bone morphogenetic protein (BMP)-Smad1/5/8 signaling alterations are emerging drivers of muscle catabolism, hence, characterizing these perturbations is pivotal to develop therapeutic approaches. We identified two promoters of "BMP resistance" in cancer cachexia, specifically the BMP scavenger erythroferrone (ERFE) and the intracellular inhibitor FKBP12. ERFE is upregulated in cachectic cancer patients' muscle biopsies and in murine cachexia models, where its expression is driven by STAT3. Moreover, the knock down of Erfe or Fkbp12 reduces muscle wasting in cachectic mice. To bypass the BMP resistance mediated by ERFE and release the brake on the signaling, we targeted FKBP12 with low-dose FK506. FK506 restores BMP-Smad1/5/8 signaling, rescuing myotube atrophy by inducing protein synthesis. In cachectic tumor-bearing mice, FK506 prevents muscle and body weight loss and protects from neuromuscular junction alteration, suggesting therapeutic potential for targeting the ERFE-FKBP12 axis.


Subject(s)
Cachexia , Neoplasms , Humans , Mice , Animals , Cachexia/drug therapy , Cachexia/etiology , Cachexia/metabolism , Tacrolimus/metabolism , Tacrolimus/pharmacology , Muscle, Skeletal/metabolism , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Neoplasms/pathology
2.
Philos Trans R Soc Lond B Biol Sci ; 378(1879): 20220169, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37122219

ABSTRACT

Cardiac ryanodine receptors (RyR2) release the Ca2+ from intracellular stores that is essential for cardiac myocyte contraction. The ion channel opening is tightly regulated by intracellular factors, including the FK506 binding proteins, FKBP12 and FKBP12.6. The impact of these proteins on RyR2 activity and cardiac contraction is debated, with often apparently contradictory experimental results, particularly for FKBP12. The isoform that regulates RyR2 has generally been considered to be FKBP12.6, despite the fact that FKBP12 is the major isoform associated with RyR2 in some species and is bound in similar proportions to FKBP12.6 in others, including sheep and humans. Here, we show time- and concentration-dependent effects of adding FKBP12 to RyR2 channels that were partly depleted of FKBP12/12.6 during isolation. The added FKBP12 displaced most remaining endogenous FKBP12/12.6. The results suggest that FKBP12 activates RyR2 with high affinity and inhibits RyR2 with lower affinity, consistent with a model of negative cooperativity in FKBP12 binding to each of the four subunits in the RyR tetramer. The easy dissociation of some FKBP12/12.6 could dynamically alter RyR2 activity in response to changes in in vivo regulatory factors, indicating a significant role for FKBP12/12.6 in Ca2+ signalling and cardiac function in healthy and diseased hearts. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.


Subject(s)
Ryanodine Receptor Calcium Release Channel , Tacrolimus Binding Protein 1A , Humans , Animals , Sheep , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Myocardium/metabolism , Calcium Signaling , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Calcium/metabolism
3.
Transplantation ; 107(2): 382-391, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36070572

ABSTRACT

BACKGROUND: . Transplant recipients may develop rejection despite having adequate tacrolimus whole blood predose concentrations (C 0 ). The intra-immune cellular concentration is potentially a better target than C 0 . However, little is known regarding intracellular tacrolimus concentration in T-lymphocytes and monocytes. We investigated the tacrolimus concentrations in both cell types and their relation with the expression and activity of FK-binding protein (FKBP)-12 and P-glycoprotein (P-gp). METHODS: . T-lymphocytes and monocytes were isolated from kidney transplant recipients followed by intracellular tacrolimus concentration measurement. FKBP-12 and P-gp were quantified with Western blot, flow cytometry, and the Rhodamine-123 assay. Interleukin-2 and interferon-γ in T-lymphocytes were measured to quantify the effect of tacrolimus. RESULTS: . Tacrolimus concentration in T-lymphocytes was lower than in monocytes (15.3 [8.5-33.4] versus 131.0 [73.5-225.1] pg/million cells; P < 0.001). The activity of P-gp (measured by Rhodamine-123 assay) was higher in T-lymphocytes than in monocytes. Flow cytometry demonstrated a higher expression of P-gp (normalized mean fluorescence intensity 1.5 [1.2-1.7] versus 1.2 [1.1-1.4]; P = 0.012) and a lower expression of FKBP-12 (normalized mean fluorescence intensity 1.3 [1.2-1.7] versus 1.5 [1.4-2.0]; P = 0.011) in T-lymphocytes than monocytes. Western blot confirmed these observations. The addition of verapamil, a P-gp inhibitor, resulted in a 2-fold higher intra-T-cell tacrolimus concentration. This was accompanied by a significantly fewer cytokine-producing cells. CONCLUSIONS: . T-lymphocytes have a higher activity of P-gp and lower concentration of the FKBP-12 compared with monocytes. This explains the relatively lower tacrolimus concentration in T-lymphocytes. The addition of verapamil prevents loss of intracellular tacrolimus during the cell isolation process and is required to ensure adequate intracellular concentration measurement.


Subject(s)
Kidney Transplantation , Tacrolimus , Humans , Tacrolimus/pharmacology , Immunosuppressive Agents/pharmacology , T-Lymphocytes/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacology , Kidney Transplantation/adverse effects , Kidney Transplantation/methods , Monocytes/metabolism , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Transplant Recipients , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/pharmacology , Verapamil/pharmacology , Rhodamines/metabolism , Rhodamines/pharmacology
4.
Elife ; 92020 01 09.
Article in English | MEDLINE | ID: mdl-31916935

ABSTRACT

The effects of the immunophilins, FKBP12 and FKBP12.6, and phosphorylation on type II ryanodine receptor (RyR2) arrangement and function were examined using correlation microscopy (line scan confocal imaging of Ca2+ sparks and dual-tilt electron tomography) and dSTORM imaging of permeabilized Wistar rat ventricular myocytes. Saturating concentrations (10 µmol/L) of either FKBP12 or 12.6 significantly reduced the frequency, spread, amplitude and Ca2+ spark mass relative to control, while the tomograms revealed both proteins shifted the tetramers into a largely side-by-side configuration. Phosphorylation of immunophilin-saturated RyR2 resulted in structural and functional changes largely comparable to phosphorylation alone. dSTORM images of myocyte surfaces demonstrated that both FKBP12 and 12.6 significantly reduced RyR2 cluster sizes, while phosphorylation, even of immunophilin-saturated RyR2, increased them. We conclude that both RyR2 cluster size and the arrangement of tetramers within clusters is dynamic and respond to changes in the cellular environment. Further, these changes affect Ca2+ spark formation.


Subject(s)
Myocytes, Cardiac/metabolism , Protein Processing, Post-Translational , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Heart Ventricles , Phosphorylation , Protein Structure, Quaternary , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum/metabolism , Tacrolimus Binding Protein 1A/pharmacology , Tacrolimus Binding Proteins/pharmacology
5.
J Gen Physiol ; 151(4): 567-577, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30635368

ABSTRACT

Effective practices to improve skeletal muscle fatigue resistance are crucial for athletes as well as patients with dysfunctional muscles. To this end, it is important to identify the cellular signaling pathway that triggers mitochondrial biogenesis and thereby increases oxidative capacity and fatigue resistance in skeletal muscle fibers. Here, we test the hypothesis that the stress induced in skeletal muscle fibers by endurance exercise causes a reduction in the association of FK506-binding protein 12 (FKBP12) with ryanodine receptor 1 (RYR1). This will result in a mild Ca2+ leak from the sarcoplasmic reticulum (SR), which could trigger mitochondrial biogenesis and improved fatigue resistance. After giving mice access to an in-cage running wheel for three weeks, we observed decreased FKBP12 association to RYR1, increased baseline [Ca2+]i, and signaling associated with greater mitochondrial biogenesis in muscle, including PGC1α1. After six weeks of voluntary running, FKBP12 association is normalized, baseline [Ca2+]i returned to values below that of nonrunning controls, and signaling for increased mitochondrial biogenesis was no longer present. The adaptations toward improved endurance exercise performance that were observed with training could be mimicked by pharmacological agents that destabilize RYR1 and thereby induce a modest Ca2+ leak. We conclude that a mild RYR1 SR Ca2+ leak is a key trigger for the signaling pathway that increases muscle fatigue resistance.


Subject(s)
Calcium/metabolism , Muscle Fatigue/physiology , Sarcoplasmic Reticulum/physiology , Animals , Anti-Bacterial Agents/pharmacology , Male , Mice , Motor Activity , Muscle, Skeletal , Protein Stability , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction , Sirolimus/pharmacology , Tacrolimus Binding Protein 1A/pharmacology
6.
Article in English | MEDLINE | ID: mdl-30181374

ABSTRACT

FK506 (tacrolimus) is an FDA-approved immunosuppressant indicated for the prevention of allograft rejections in patients undergoing organ transplants. In mammals, FK506 inhibits the calcineurin-nuclear factor of activated T cells (NFAT) pathway to prevent T-cell proliferation by forming a ternary complex with its binding protein, FKBP12, and calcineurin. FK506 also exerts antifungal activity by inhibiting calcineurin, which is essential for the virulence of human-pathogenic fungi. Nevertheless, FK506 cannot be used directly as an antifungal drug due to its immunosuppressive action. In this study, we analyzed the cytotoxicity, immunosuppressive activity, and antifungal activity of four FK506 analogs, 31-O-demethyl-FK506, 9-deoxo-FK506, 9-deoxo-31-O-demethyl-FK506, and 9-deoxo-prolyl-FK506, in comparison with that of FK506. The four FK506 analogs generally possessed lower cytotoxicity and immunosuppressive activity than FK506. The FK506 analogs, except for 9-deoxo-prolyl-FK506, had strong antifungal activity against Cryptococcus neoformans and Candida albicans, which are two major invasive pathogenic yeasts, due to the inhibition of the calcineurin pathway. Furthermore, the FK506 analogs, except for 9-deoxo-prolyl-FK506, had strong antifungal activity against the invasive filamentous fungus Aspergillus fumigatus Notably, 9-deoxo-31-O-demethyl-FK506 and 31-O-demethyl-FK506 exhibited robust synergistic antifungal activity with fluconazole, similar to FK506. Considering the antifungal efficacy, cytotoxicity, immunosuppressive activity, and synergistic effect with commercial antifungal drugs, we selected 9-deoxo-31-O-demethyl-FK506 for further evaluation of its in vivo antifungal efficacy in a murine model of systemic cryptococcosis. Although 9-deoxo-31-O-demethyl-FK506 alone was not sufficient to treat the cryptococcal infection, when it was used in combination with fluconazole, it significantly extended the survival of C. neoformans-infected mice, confirming the synergistic in vivo antifungal efficacy between these two agents.


Subject(s)
Antifungal Agents/pharmacology , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Animals , Aspergillosis/drug therapy , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Calcineurin/pharmacology , Calcineurin Inhibitors/pharmacology , Candida albicans/drug effects , Candidiasis/drug therapy , Candidiasis/microbiology , Cells, Cultured , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Female , Fluconazole/pharmacology , Immunosuppressive Agents/pharmacology , Male , Mice , Microbial Sensitivity Tests/methods , Tacrolimus Binding Protein 1A/pharmacology
7.
Nature ; 497(7448): 217-23, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23636326

ABSTRACT

The mammalian target of rapamycin (mTOR), a phosphoinositide 3-kinase-related protein kinase, controls cell growth in response to nutrients and growth factors and is frequently deregulated in cancer. Here we report co-crystal structures of a complex of truncated mTOR and mammalian lethal with SEC13 protein 8 (mLST8) with an ATP transition state mimic and with ATP-site inhibitors. The structures reveal an intrinsically active kinase conformation, with catalytic residues and a catalytic mechanism remarkably similar to canonical protein kinases. The active site is highly recessed owing to the FKBP12-rapamycin-binding (FRB) domain and an inhibitory helix protruding from the catalytic cleft. mTOR-activating mutations map to the structural framework that holds these elements in place, indicating that the kinase is controlled by restricted access. In vitro biochemistry shows that the FRB domain acts as a gatekeeper, with its rapamycin-binding site interacting with substrates to grant them access to the restricted active site. Rapamycin-FKBP12 inhibits the kinase by directly blocking substrate recruitment and by further restricting active-site access. The structures also reveal active-site residues and conformational changes that underlie inhibitor potency and specificity.


Subject(s)
TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Catalytic Domain/drug effects , Crystallography, X-Ray , Furans/chemistry , Furans/pharmacology , Humans , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Magnesium/chemistry , Magnesium/metabolism , Models, Molecular , Naphthyridines/chemistry , Naphthyridines/metabolism , Naphthyridines/pharmacology , Protein Structure, Tertiary/drug effects , Purines/chemistry , Purines/metabolism , Purines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/chemistry , Sirolimus/metabolism , Sirolimus/pharmacology , Structure-Activity Relationship , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology , mTOR Associated Protein, LST8 Homolog
8.
Methods Mol Biol ; 821: 15-28, 2012.
Article in English | MEDLINE | ID: mdl-22125057

ABSTRACT

The mammalian target of rapamycin (mTOR) is the catalytic subunit of two multiprotein complexes, mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2). Clinically used rapamycin and rapalogs are FKBP12-dependent allosteric inhibitors of mTORC1. The recently discovered WYE-125132 and related drugs represent a new generation of ATP competitive and highly specific inhibitors targeting mTOR globally. As mTORC1 and mTORC2 mediate diverse sets of both redundant and distinctive cellular pathways of growth, nutrient and energy homeostasis, rapamycin and WYE-125132 elicit both overlapping and distinctive pharmacological properties with important implications in treating cancer, metabolic, and age-related degenerative diseases. Detailed methods are described for the determination of mTOR inhibition by rapamycin and WYE-125132 in assays of recombinant mTOR enzyme, immunprecipitated native mTOR complexes, growth factor- and amino acid-induced cellular phosphorylation cascades as well as the PI3K/AKT/mTOR hyperactive breast tumor model in vitro and in vivo. The methods have been particularly useful in discovery and biochemical characterization of mTOR inhibitors, cellular and in vivo mTOR substrate phosphorylation analysis, and in deciphering novel biomarkers of mTORC1 and mTORC2 signaling pathways.


Subject(s)
Drug Discovery/methods , Protein Kinase Inhibitors/chemistry , Proteins/antagonists & inhibitors , Sirolimus/pharmacology , Transcription Factors/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Biomarkers, Tumor/antagonists & inhibitors , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Pyrazoles/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases , Tacrolimus Binding Protein 1A/pharmacology , Transcription Factors/metabolism
9.
Biochemistry ; 49(43): 9345-52, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20828147

ABSTRACT

The neuronal protein α-synuclein (α-syn) plays a central role in Parkinson's disease (PD). The pathological features of PD are the loss of dopaminergic neurons in the substantia nigra pars compacta and the presence of Lewy bodies. The C-terminal domain of α-syn is characterized by the presence of 15 acidic amino acids and all five proline residues of the protein (P108, P117, P120, P128, and P138). The aggregation of this natively unfolded protein is accelerated in vitro by FK506 binding proteins (FKBPs) showing peptidyl-prolyl cis-trans isomerase activity. These proteins catalyze the cis-trans conformational change of the X-Pro peptide bond, often a rate-limiting step in protein folding. The acceleration of the folding of α-syn by FKBPs may accelerate disease-associated aggregation. To further elucidate the role of the proline residues in the conformation and aggregation of α-syn, we constructed several mutants of α-syn in which one or more proline residues are mutated to alanine via site-directed mutagenesis. For this purpose, we produced and purified His-WT α-syn, a recombinant α-syn with a polyhistidine tag (six His residues) and a linker, and a number of Pro-to-Ala mutants. The aggregation kinetics of these mutants and His-WT α-syn were studied by turbidity, thioflavin T fluorescence, and CD measurements. We can conclude that mutation of the proline residues to alanine accelerates the aggregation kinetics of α-syn while all proline mutants formed fibrils similar to His-WT α-syn, as visualized via transmission electron microscopy. We also demonstrate that the accelerating effect of hFKBP12 is abolished via removal of the proline residues from the C-terminus. Finally, we show that the mutant of His α-syn with all five proline residues mutated to alanine is more structured (more α-helix) than His-WT α-syn, indicating the role of the Pro residues as potential helix breakers in the inhibitory conformation of the C-terminus.


Subject(s)
Proline/chemistry , Protein Multimerization , alpha-Synuclein/chemistry , Humans , Kinetics , Mutagenesis, Site-Directed , Protein Conformation , Tacrolimus Binding Protein 1A/pharmacology , alpha-Synuclein/genetics
10.
Cardiovasc Res ; 85(1): 68-78, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19661110

ABSTRACT

AIMS: Our objective was to explore the functional interdependence of protein kinase A (PKA) phosphorylation with binding of modulatory FK506 binding proteins (FKBP12/12.6) to the ryanodine receptor (RyR). RyR type 1 or type 2 was prepared from rabbit skeletal muscle or pig cardiac muscle, respectively. In heart failure, RyR2 dysfunction is implicated in fatal arrhythmia and RyR1 dysfunction is associated with muscle fatigue. A controversial underlying mechanism of RyR1/2 dysfunction is proposed to be hyperphosphorylation of RyR1/2 by PKA, causing loss of FKBP12/12.6 binding that is reversible by the experimental inhibitory drug K201 (JTV519). Phosphorylation is also a trigger for fatal arrhythmia in catecholaminergic polymorphic ventricular tachycardia associated with point mutations in RyR2. METHODS AND RESULTS: Equilibrium binding kinetics of RyR1/2 to FKBP12/12.6 were measured using surface plasmon resonance (Biacore). Free Ca(2+) concentration was used to modulate the open/closed conformation of RyR1/2 channels measured using [(3)H]ryanodine binding assays. The affinity constant-K(A), for RyR1/2 binding to FKBP12/12.6, was significantly greater for the closed compared with the open conformation. The effect of phosphorylation or K201 was to reduce the K(A) of the closed conformation by increasing the rate of dissociation k(d). K201 reduced [(3)H]ryanodine binding to RyR1/2 at all free Ca(2+) concentrations including PKA phosphorylated preparations. CONCLUSION: The results are explained through a model proposing that phosphorylation and K201 acted similarly to change the conformation of RyR1/2 and regulate FKBP12/12.6 binding. K201 stabilized the conformation, whereas phosphorylation facilitated a subsequent molecular event that might increase the rate of an open/closed conformational transition.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Proteins/metabolism , Thiazepines/pharmacology , Animals , Male , Phosphorylation , Protein Conformation , Rabbits , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Swine , Tacrolimus Binding Protein 1A/pharmacology
11.
J Neurochem ; 106(1): 121-33, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18346205

ABSTRACT

Aggregation of alpha-synuclein (alpha-SYN) plays a key role in Parkinson's disease. We have previously shown that aggregation of alpha-SYN in vitro is accelerated by addition of FK506 binding proteins (FKBP) and that this effect can be counteracted by FK506, a specific inhibitor of these enzymes. In this paper, we investigated in detail the effect of FKBP12 on early aggregation and on fibril formation of wild-type, A53T and A30P alpha-SYN. FKBP12 has a much smaller effect on the fibril formation of these two clinical mutants alpha-SYN. Using an inactive enzyme, we were able to discriminate between catalytic and non-catalytic effects that differentially influence the two processes. A model explaining non-linear concentration dependencies is proposed.


Subject(s)
Brain/metabolism , Nerve Degeneration/metabolism , Neurofibrillary Tangles/metabolism , Neurons/metabolism , Tacrolimus Binding Protein 1A/metabolism , alpha-Synuclein/metabolism , Amino Acid Substitution , Brain/pathology , Brain/physiopathology , Catalytic Domain/genetics , Cell Line, Tumor , Humans , Immunosuppressive Agents/pharmacology , Mutation/genetics , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/genetics , Neurons/drug effects , Neurons/pathology , Nonlinear Dynamics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/pharmacology , Time Factors , alpha-Synuclein/drug effects , alpha-Synuclein/genetics
12.
FASEB J ; 20(3): 524-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16410343

ABSTRACT

Aggregation of alpha-synuclein (alpha-SYN) plays a key role in Parkinson's disease (PD). We have used fluorescence correlation spectroscopy (FCS) to study alpha-SYN aggregation in vitro and discovered that this process is clearly accelerated by addition of FK506 binding proteins (FKBPs). This effect was observed both with E. coli SlyD FKBP and with human FKBP12 and was counteracted by FK506, a specific inhibitor of FKBP. The alpha-SYN aggregates formed in the presence of FKBP12 showed fibrillar morphology. The rotamase activity of FKBP apparently accelerates the folding and subsequent aggregation of alpha-SYN. Since FK506 and other non-immunosuppressive FKBP inhibitors are known to display neuroregenerative and neuroprotective properties in disease models, the observed inhibition of rotamase activity and alpha-SYN aggregation, may explain their mode of action. Our results open perspectives for the treatment of PD with immunophilin ligands that inhibit a specific member of the FKBP family.


Subject(s)
Escherichia coli Proteins/pharmacology , Peptidylprolyl Isomerase/pharmacology , Tacrolimus Binding Protein 1A/pharmacology , alpha-Synuclein/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/physiology , Humans , Microscopy, Electron , Nephelometry and Turbidimetry , Peptidylprolyl Isomerase/isolation & purification , Peptidylprolyl Isomerase/physiology , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Spectrometry, Fluorescence , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/physiology , alpha-Synuclein/drug effects , alpha-Synuclein/genetics , alpha-Synuclein/ultrastructure
13.
J Cell Sci ; 118(Pt 23): 5443-51, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16278292

ABSTRACT

Ca2+ release from the sarcoplasmic reticulum (SR) by the IP3 receptors (IP3Rs) crucially regulates diverse cell signalling processes from reproduction to apoptosis. Release from the IP3R may be modulated by endogenous proteins associated with the receptor, such as the 12 kDa FK506-binding protein (FKBP12), either directly or indirectly by inhibition of the phosphatase calcineurin. Here, we report that, in addition to calcineurin, FKPBs modulate release through the mammalian target of rapamycin (mTOR), a kinase that potentiates Ca2+ release from the IP3R in smooth muscle. The presence of FKBP12 was confirmed in colonic myocytes and co-immunoprecipitated with the IP3R. In aortic smooth muscle, however, although present, FKBP12 did not co-immunoprecipitate with IP3R. In voltage-clamped single colonic myocytes rapamycin, which together with FKBP12 inhibits mTOR (but not calcineurin), decreased the rise in cytosolic Ca2+ concentration ([Ca2+]c) evoked by IP3R activation (by photolysis of caged IP3), without decreasing the SR luminal Ca2+ concentration ([Ca2+]l) as did the mTOR inhibitors RAD001 and LY294002. However, FK506, which with FKBP12 inhibits calcineurin (but not mTOR), potentiated the IP3-evoked [Ca2+]c increase. This potentiation was due to the inhibition of calcineurin; it was mimicked by the phosphatase inhibitors cypermethrin and okadaic acid. The latter two inhibitors also prevented the FK506-evoked increase as did a calcineurin inhibitory peptide (CiP). In aortic smooth muscle, where FKBP12 was not associated with IP3R, the IP3-mediated Ca2+ release was unaffected by FK506 or rapamycin. Together, these results suggest that FKBP12 has little direct effect on IP3-mediated Ca2+ release, even though it is associated with IP3R in colonic myocytes. However, FKBP12 might indirectly modulate Ca2+ release through two effector proteins: (1) mTOR, which potentiates and (2) calcineurin, which inhibits Ca2+ release from IP3R in smooth muscle.


Subject(s)
Calcineurin/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Calcium Channels/drug effects , Cells, Cultured , Chromones/pharmacology , Everolimus , Guinea Pigs , Inositol 1,4,5-Trisphosphate Receptors , Male , Morpholines/pharmacology , Muscle, Smooth, Vascular/cytology , Patch-Clamp Techniques , Protein Kinases/drug effects , Receptors, Cytoplasmic and Nuclear/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology
14.
Eur J Pharmacol ; 509(1): 11-9, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15713424

ABSTRACT

Immunophilin ligands are neuroregenerative agents, characterized by binding to FK506 binding proteins (FKBPs), which stimulate recovery of neurons in a variety of injury paradigms. Here we report the discovery of a novel, non-immunosuppressive immunophilin ligand, FK1706. FK1706, a derivative of FK506, showed similarly high affinity for two FKBP subtypes, FKBP-12 and FKBP-52, but inhibited T-cell proliferation and interleukin-2 cytokine production with much lower potency and efficacy than FK506. FK1706 (0.1 to 10 nM) significantly potentiated nerve growth factor (NGF)-induced neurite outgrowth in SH-SY5Y cells, as did FK506. This neurite potentiation could be blocked by an anti-FKBP-52 antibody, as well as by specific pharmacological inhibitors of phospholipase C (PLC), phosphatidylinositol 3-kinase (PI3K), and the Ras/Raf/Mitogen-Activated Protein Kinase (MAPK) signaling pathway. FK1706 also potentiated NGF-induced MAPK activation, with a similar dose-dependency to that necessary for potentiating neurite outgrowth. Taken together, these data suggest that FK1706 is a non-immunosuppressive immunophilin ligand with significant neurotrophic effects, putatively mediated via FKBP-52 and the Ras/Raf/MAPK signaling pathway, and therefore that FK1706 may have therapeutic potential in a variety of neurological disorders.


Subject(s)
Immunophilins/pharmacology , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Immunophilins/chemistry , Immunophilins/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/antagonists & inhibitors , Receptor, trkB/metabolism , Signal Transduction/physiology , Tacrolimus/analogs & derivatives , Tacrolimus/chemistry , Tacrolimus/immunology , Tacrolimus/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/pharmacology , Tritium
15.
Am J Physiol Cell Physiol ; 287(1): C36-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14985235

ABSTRACT

We showed that frog alpha-ryanodine receptor (alpha-RyR) had a lower gain of Ca(2+)-induced Ca(2+) release (CICR) activity than beta-RyR in sarcoplasmic reticulum (SR) vesicles, indicating selective "stabilization" of the former isoform (Murayama T and Ogawa Y. J Biol Chem 276: 2953-2960, 2001). To know whether this is also the case with mammalian RyR1, we determined [(3)H]ryanodine binding of RyR1 and RyR3 in bovine diaphragm SR vesicles. The value of [(3)H]ryanodine binding (B) was normalized by the number of maximal binding sites (B(max)), whereby the specific activity of each isoform was expressed. This B/B(max) expression demonstrated that ryanodine binding of individual channels for RyR1 was <15% that for RyR3. Responses to Ca(2+), Mg(2+), adenine nucleotides, and caffeine were not substantially different between in situ and purified isoforms. These results suggest that the gain of CICR activity of RyR1 is markedly lower than that of RyR3 in mammalian skeletal muscle, indicating selective stabilization of RyR1 as is true of frog alpha-RyR. The stabilization was partly eliminated by FK506 and partly by solubilization of the vesicles with CHAPS, each of which was additive to the other. In contrast, high salt, which greatly enhances [(3)H]ryanodine binding, caused only a minor effect on the stabilization of RyR1. None of the T-tubule components, coexisting RyR3, or calmodulin was the cause. The CHAPS-sensitive intra- and intermolecular interactions that are common between mammalian and frog skeletal muscles and the isoform-specific inhibition by FKBP12, which is characteristic of mammals, are likely to be the underlying mechanisms.


Subject(s)
Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Adenine Nucleotides/pharmacology , Animals , Binding Sites , Caffeine/pharmacology , Calmodulin/metabolism , Cattle , Cholic Acids/pharmacology , Detergents/pharmacology , Diaphragm/metabolism , Dose-Response Relationship, Drug , Magnesium/pharmacology , Phospholipids/pharmacology , Precipitin Tests , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/drug effects , Sodium Chloride/administration & dosage , Solubility , Tacrolimus Binding Protein 1A/pharmacology
16.
Arch Virol ; 147(8): 1531-42, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12181673

ABSTRACT

Peptides of the V3 loop of the HIV-1 envelope glycoprotein gp120 have been shown to bind with high affinity to the immunophilins cyclophilin (Cyp) A, CypB and the FK506-binding protein 12 (FKBP12) [10]. We investigated whether immunophilins affect HIV-1 infection by assuming they are able to bind to the V3 loop of gp120. T cells and peripheral blood mononuclear cells were infected with T-cell-tropic or macrophage-tropic HIV-1 strains, respectively, in the presence of different concentrations of immunophilins. P24 antigen ELISA and real-time PCR measurements demonstrated that exogenously added immunophilins do not influence HIV-1 infection. CypA is known to interact with the HIV-1 Gag polyprotein and to be incorporated into the virions. This incorporation can be prevented by cyclosporin A (CsA) resulting in a decreased yield of infectious virus, the mechanism of which is unknown. We measured a normal production of proviral DNA in the first round of infection in CsA treated cells but afterwards, infection was decreased if CsA was present. Pre-treatment of the HIV-1 inocula with CsA, blocking the function of virus-associated CypA, did not inhibit the ensuing yield of infection. We therefore may conclude that endogenous CypA exerts its action after reverse transcription but before virus maturation, probably during capsid formation. FK520, an immunosuppressor which binds to FKBP, had no effect on HIV-1 infection.


Subject(s)
Cyclophilin A/pharmacology , Cyclophilins/pharmacology , HIV-1/drug effects , Tacrolimus Binding Protein 1A/pharmacology , Cyclosporine/pharmacology , HIV-1/physiology , Humans , Leukocytes, Mononuclear/virology , Peptidylprolyl Isomerase , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology
17.
Eur J Biochem ; 269(14): 3540-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12135494

ABSTRACT

The calcineurin (CaN) alpha and beta catalytic subunit isoforms are coexpressed within almost all cell types. The enzymatic properties of CaN heterodimers comprised of the regulatory B subunit (CnB) with either the alpha or beta catalytic subunit were compared using in vitro phosphatase assays. CaN containing the alpha isoform (CnA alpha) has lower K(m) and higher V(max) values than CaN containing the beta isoform (CnA beta) toward the PO4-RII, PO4-DARPP-32(20-38) peptides, and p-nitrophenylphosphate (pNPP). CaN heterodimers containing the alpha or beta catalytic subunit isoform displayed identical calmodulin dissociation rates. Similar inhibition curves for each CaN heterodimer were obtained with the CaN autoinhibitory peptide (CaP) and cyclophilin A/cyclosporin A (CyPA/CsA) using each peptide substrate at K(m) concentrations, except for a five- to ninefold higher IC50 value measured for CaN containing the beta isoform with p-nitrophenylphosphate as substrate. No difference in stimulation of phosphatase activity toward p-nitrophenylphosphate by FKBP12/FK506 was observed. At low concentrations of FKBP12/FK506, CaN containing the alpha isoform is more sensitive to inhibition than CaN containing the beta isoform using the phosphopeptide substrates. Higher concentrations of FKBP12/FK506 are required for maximal inhibition of beta CaN using PO4-DARPP-32(20-38) as substrate. The functional differences conferred upon CaN by the alpha or beta catalytic subunit isoforms suggest that the alpha:beta and CaN:substrate ratios may determine the levels of CaN phosphatase activity toward specific substrates within tissues and specific cell types. These findings also indicate that the alpha and beta catalytic subunit isoforms give rise to substrate-dependent differences in sensitivity toward FKBP12/FK506.


Subject(s)
Calcineurin Inhibitors , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Tacrolimus/pharmacology , Animals , Apoptosis Regulatory Proteins , Calcineurin/chemistry , Calmodulin/metabolism , Carrier Proteins/pharmacology , Dimerization , Humans , Isoenzymes/chemistry , Nitrophenols/metabolism , Organophosphorus Compounds/metabolism , Protein Multimerization , Protein Subunits , Rats , Substrate Specificity , Tacrolimus Binding Protein 1A/pharmacology
18.
Neuroscience ; 110(2): 213-25, 2002.
Article in English | MEDLINE | ID: mdl-11958864

ABSTRACT

The Ca(2+)/calmodulin-dependent protein phosphatase, calcineurin, modulates a number of key Ca(2+) signaling pathways in neurons, and has been implicated in Ca(2+)-dependent negative feedback inactivation of N-methyl-D-aspartate receptors and voltage-sensitive Ca(2+) channels. In contrast, we report here that three mechanistically disparate calcineurin inhibitors, FK-506, cyclosporin A, and the calcineurin autoinhibitory peptide, inhibited high-voltage-activated Ca(2+) channel currents by up to 40% in cultured hippocampal neurons, suggesting that calcineurin acts to enhance Ca(2+) currents. This effect occurred with Ba(2+) or Ca(2+) as charge carrier, and with or without intracellular Ca(2+) buffered by EGTA. Ca(2+)-dependent inactivation of Ca(2+) channels was not affected by FK-506. The immunosuppressant, rapamycin, and the protein phosphatase 1/2A inhibitor, okadaic acid, did not decrease Ca(2+) channel current, showing specificity for effects on calcineurin. Blockade of L-type Ca(2+) channels with nimodipine fully negated the effect of FK-506 on Ca(2+) channel current, while blockade of N-, and P-/Q-type Ca(2+) channels enhanced FK-506-mediated inhibition of the remaining L-type-enriched current. FK-506 also inhibited substantially more Ca(2+) channel current in 4-week-old vs. 2-week-old cultures, an effect paralleled by an increase in calcineurin A mRNA levels. These studies provide the first evidence that calcineurin selectively enhances L-type Ca(2+) channel activity in neurons. Moreover, this action appears to be increased concomitantly with the well-characterized increase in L-type Ca(2+) channel availability in hippocampal neurons with age-in-culture.


Subject(s)
Aging/metabolism , Calcineurin/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Cell Differentiation/physiology , Hippocampus/growth & development , Hippocampus/metabolism , Neurons/metabolism , Animals , Apoptosis Regulatory Proteins , Calcineurin/genetics , Calcineurin Inhibitors , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Cyclosporine/pharmacology , Female , Fetus , Hippocampus/drug effects , Immunosuppressive Agents/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Pregnancy , Protein Phosphatase 1 , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/pharmacology
19.
Chem Biol ; 9(1): 49-61, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841938

ABSTRACT

We used olefin metathesis to synthesize C40 derivatives of FK506 and measured their ability, when complexed to FKBP12, to inhibit calcineurin's phosphatase activity. We identified modular dimerization domains (CABs) containing segments of the calcineurin A and B polypeptides. These CABs respond to FK506 both when overexpressed in mammalian cells and in yeast or mammalian three-hybrid assays. Using chemical genetic selection, we identified compensatory mutant CABs that respond to a calcineurin-resistant FK506 derivative at concentrations well below the response threshold for CABs containing only wild-type calcineurin sequence. These reagents provide a small molecule-protein combination orthogonal to existing dimerizer systems and may be used with existing systems to increase the complexity of induced-proximity experiments. This new use of the "bump-hole" strategy protects target cells from complications arising from the inhibition of endogenous calcineurin.


Subject(s)
Calcineurin Inhibitors , Calcineurin/chemistry , Enzyme Inhibitors/chemistry , Immunosuppressive Agents/chemistry , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus/chemistry , Dimerization , Enzyme Inhibitors/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Jurkat Cells , Models, Molecular , Mutation , Recombinant Fusion Proteins , Saccharomyces cerevisiae/genetics , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/pharmacology
20.
J Biol Chem ; 276(49): 46533-43, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11571290

ABSTRACT

The bone morphogenetic proteins (BMPs) regulate early embryogenesis and morphogenesis of multiple organs, such as bone, kidney, limbs, and muscle. Smad1 is one of the key signal transducers of BMPs and is responsible for transducing receptor activation signals from the cytoplasm to the nucleus, where Smad1 serves as a transcriptional regulator of various BMP-responsive genes. Based upon the ability of Smad1 to bind multiple proteins involved in proteasome-mediated degradation pathway, we investigated whether Smad1 could be a substrate for proteasome. We found that Smad1 is targeted to proteasome for degradation in response to BMP type I receptor activation. The targeting of Smad1 to proteasome involves not only the receptor activation-induced Smad1 ubiquitination but also the targeting functions of the ornithine decarboxylase antizyme and the proteasome beta subunit HsN3. Our studies provide the first evidence for BMP-induced proteasomal targeting and degradation of Smad1 and also reveal new players and novel mechanisms involved in this important aspect of Smad1 regulation and function.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases , Receptors, Growth Factor , Trans-Activators/metabolism , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Proteins/physiology , Humans , Hydrolysis , Mice , Proteasome Endopeptidase Complex , Smad Proteins , Smad1 Protein , Tacrolimus Binding Protein 1A/pharmacology , Tumor Cells, Cultured , Two-Hybrid System Techniques , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...